Acessibilidade / Reportar erro

Extracellular vesicles in Alzheimer's disease

Vesículas extracelulares na doença de Alzheimer

Abstract

Extracellular vesicles (EVs) are small vesicles released by cells that facilitate cell signaling. They are categorized based on their biogenesis and size. In the context of the central nervous system (CNS), EVs have been extensively studied for their role in both normal physiological functions and diseases like Alzheimer's disease (AD). AD is a neurodegenerative disorder characterized by cognitive decline and neuronal death. EVs have emerged as potential biomarkers for AD due to their involvement in disease progression. Specifically, EVs derived from neurons, astrocytes, and neuron precursor cells exhibit changes in quantity and composition in AD. Neuron-derived EVs have been found to contain key proteins associated with AD pathology, such as amyloid beta (Aß) and tau. Increased levels of Aß in neuron-derived EVs isolated from the plasma have been observed in individuals with AD and mild cognitive impairment, suggesting their potential as early biomarkers. However, the analysis of tau in neuron-derived EVs is still inconclusive. In addition to Aß and tau, neuron-derived EVs also carry other proteins linked to AD, including synaptic proteins. These findings indicate that EVs could serve as biomarkers for AD, particularly for early diagnosis and disease monitoring. However, further research is required to validate their use and explore potential therapeutic applications. To summarize, EVs are small vesicles involved in cell signaling within the CNS. They hold promise as biomarkers for AD, potentially enabling early diagnosis and monitoring of disease progression. Ongoing research aims to refine their use as biomarkers and uncover additional therapeutic applications.

Keywords
Extracellular Vesicles; Central Nervous System; Alzheimer Disease; Biomarkers; Early Diagnosis; Signal Transduction

Resumo

As vesículas extracelulares (VEs) são pequenas estruturas liberadas pelas células que agem na sinalização celular. No sistema nervoso central (SNC), as VEs são estudadas em relação à doença de Alzheimer (DA), um distúrbio neurodegenerativo que cursa com declínio cognitivo e morte neuronal. As VEs podem ser biomarcadores potenciais para a DA devido ao seu papel na progressão da doença. As VEs derivadas de neurônios, astrócitos e células precursoras apresentam alterações na DA, contendo proteínas associadas à patologia da DA, como beta-amiloide (Aß) e tau. Níveis elevados de Aß foram observados nas VEs de neurônios de indivíduos com DA, sugerindo seu potencial como biomarcadores precoces. A análise de tau nas VEs de neurônios ainda é inconclusiva. Além disso, as VEs neurais carregam outras proteínas relacionadas à DA, incluindo proteínas sinápticas. As VEs podem ser promissoras como biomarcadores para o diagnóstico precoce e monitoramento da DA, porém mais pesquisas são necessárias para validar seu uso e explorar aplicações terapêuticas. Em resumo, as VEs são vesículas envolvidas na sinalização celular no SNC, com potencial como biomarcadores para a DA.

Palavras-chave
Vesículas Extracelulares; Sistema Nervoso Central; Doença de Alzheimer; Biomarcadores; Diagnóstico Precoce; Transdução de Sinais

INTRODUCTION

Extracellular vesicles (EVs) are defined as vesicles bounded by a phospholipid bilayer originating from endosomes or from the evagination of the plasma membrane that do not have the ability to replicate itself.11 Théry C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018;7(01):1535750. Doi: 10.1080/20013078.2018.1535750
https://doi.org/10.1080/20013078.2018.15...
EVs can be classified according to different parameters, such as the mechanism of biogenesis or with respect to their size, even if the latter method of differentiation is not very effective, since an overlap between size ranges of different types of extracellular vesicles occurs.11 Théry C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018;7(01):1535750. Doi: 10.1080/20013078.2018.1535750
https://doi.org/10.1080/20013078.2018.15...

2 Théry C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol 2009;9(08):581–593. Doi: 10.1038/nri2567
https://doi.org/10.1038/nri2567...

3 Denzer K, Kleijmeer MJ, Heijnen HF, Stoorvogel W, Geuze HJ. Exosome: from internal vesicle of the multivesicular body to intercellular signaling device. J Cell Sci 2000;113(Pt 19): 3365–3374. Doi: 10.1242/jcs.113.19.3365
https://doi.org/10.1242/jcs.113.19.3365...
-44 György B, Szabó TG, Pásztói M, et al. Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles. Cell Mol Life Sci 2011;68(16):2667–2688. Doi: 10.1007/s00018-011-0689-3
https://doi.org/10.1007/s00018-011-0689-...
Thus, recommendations from the International Society of Extracellular Vesicles suggest, when the origin is unclear, identifying ~100-1000 nm vesicles as "large EVs" (lEVs) and ~40-100 nm vesicles as "small EVs" (sEVs).

Regarding their biogenesis, EVs can be formed from the evagination of the plasma membrane, to form the so-called microvesicles (MVs) or microparticles (MPs), or they can be formed from the invagination of the membrane of endosomes, forming multivesicular bodies (MVBs) that contain in their interior exosomes, which are released from the fusion of the MVB with the plasma membrane.22 Théry C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol 2009;9(08):581–593. Doi: 10.1038/nri2567
https://doi.org/10.1038/nri2567...
,33 Denzer K, Kleijmeer MJ, Heijnen HF, Stoorvogel W, Geuze HJ. Exosome: from internal vesicle of the multivesicular body to intercellular signaling device. J Cell Sci 2000;113(Pt 19): 3365–3374. Doi: 10.1242/jcs.113.19.3365
https://doi.org/10.1242/jcs.113.19.3365...
Once in the extracellular millieu, EVs act as important cell signaling mediators since they carry several important molecules, such as proteins, lipids, carbohydrates, and genetic material.55 Raposo G, StoorvogelW. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 2013;200(04):373–383. Doi: 10.1083/jcb.201211138
https://doi.org/10.1083/jcb.201211138...
,66 Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function.Nat Rev Immunol 2002;2(08):569–579. Doi: 10.1038/nri855
https://doi.org/10.1038/nri855...
In this sense, one can affirm that EVs may be involved with pathological processes and even serve as biomarkers of various diseases. In this review, we summarize the accumulating evidence that supports the idea of using EVs as biomarkers that could enable not only the diagnosis but also monitoring the progression of Alzheimer's disease.

EXTRACELLULAR VESICLES IN CELLULAR COMMUNICATION, CENTRAL NERVOUS SYSTEM PHYSIOLOGY AND ITS ROLE ON THE PATHOPHYSIOLOGY OF DISEASES

EVs can be found in various biofluids, not only in blood, and their content may reflect both their origin and function.77 Shah R, Patel T, Freedman JE. Circulating Extracellular Vesicles in Human Disease. N Engl J Med 2018;379(10):958–966. Doi: 10.1056/NEJMra1704286
https://doi.org/10.1056/NEJMra1704286...
Regarding function, it is thought that EVs may serve as a cell signaling mechanism, being involved in systemic processes, such as immune response and inflammation, as well as pathological processes, which has increased the interest in their potential as biomarkers of such diseases.88 Raposo G, Nijman HW, StoorvogelW, et al. B lymphocytes secrete antigen-presenting vesicles. J Exp Med 1996;183(03):1161–1172,99 Robbins PD, Dorronsoro A, Booker CN. Regulation of chronic inflammatory and immune processes by extracellular vesicles. J Clin Invest 2016;126(04):1173–1180 Colombo et al. 20141010 Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol 2014;30:255–289. Doi: 10.1146/annurevcellbio-101512-122326
https://doi.org/10.1146/annurev-cellbio-...
demonstrated the participation of exosomes in processes such as angiogenesis, inflammation, transport of morphogens, and apoptosis. The EV release by central nervous system (CNS) cells, such as neurons, astrocytes, oligodendrocytes, and microglia, has been established for many years.1111 Fauré J, Lachenal G, Court M, et al. Exosomes are released by cultured cortical neurones. Mol Cell Neurosci 2006;31(04): 642–648. Doi: 10.1016/j.mcn.2005.12.003
https://doi.org/10.1016/j.mcn.2005.12.00...

12 Morel L, Regan M, Higashimori H, et al. Neuronal exosomal miRNA-dependent translational regulation of astroglial glutamate transporter GLT1. J Biol Chem 2013;288(10):7105–7116. Doi: 10.1074/jbc.M112.410944
https://doi.org/10.1074/jbc.M112.410944...

13 Potolicchio I, Carven GJ, Xu X, et al. Proteomic analysis of microglia-derived exosomes: metabolic role of the aminopeptidase CD13 in neuropeptide catabolism. J Immunol 2005;175(04): 2237–2243. Doi: 10.4049/jimmunol.175.4.2237
https://doi.org/10.4049/jimmunol.175.4.2...
-1414 Krämer-Albers EM, Bretz N, Tenzer S, et al. Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: Trophic support for axons? Proteomics Clin Appl 2007;1 (11):1446–1461. Doi: 10.1002/prca.200700522
https://doi.org/10.1002/prca.200700522...
There are several studies that investigated some of the physiological actions that EVs play in the central nervous system, although most of them are in vitro or in animal models. It is established that active neurons secrete exosomes containing various contents, mostly proteins and genetic material, which act by altering the expression of proteins in the recipient neurons, as well as by modulating neurotransmission and even neurogenesis.1515 Saeedi S, Israel S, Nagy C, Turecki G. The emerging role of exosomes in mental disorders. Transl Psychiatry 2019;9(01): 122. Doi: 10.1038/s41398-019-0459-9
https://doi.org/10.1038/s41398-019-0459-...
Moreover, other studies have already demonstrated that the internalization of EVs secreted by certain groups of neurons, by other groups of neurons, may affect the process of synaptic plasticity.1616 Chivet M, Javalet C, Hemming F, et al. Exosomes as a novel way of interneuronal communication. Biochem Soc Trans 2013;41(01): 241–244. Doi: 10.1042/BST20120266
https://doi.org/10.1042/BST20120266...
,1717 Goldie BJ, Dun MD, Lin M, et al. Activity-associated miRNA are packaged in Map1b-enriched exosomes released from depolarized neurons. Nucleic Acids Res 2014;42(14):9195–9208. Doi: 10.1093/nar/gku594
https://doi.org/10.1093/nar/gku594...
Neurons can also communicate with glial cells via EVs. For example, one study demonstrated that neuron-derived exosomes containing a family of miRNAs, when internalized by astrocytes, led to increased expression of several proteins, including glutamate-transporter-1(GLT-1). This protein plays a key role in maintaining the homeostasis of glutamatergic synapses.1212 Morel L, Regan M, Higashimori H, et al. Neuronal exosomal miRNA-dependent translational regulation of astroglial glutamate transporter GLT1. J Biol Chem 2013;288(10):7105–7116. Doi: 10.1074/jbc.M112.410944
https://doi.org/10.1074/jbc.M112.410944...
,1818 Yang Y, Gozen O, Vidensky S, RobinsonMB, Rothstein JD. Epigenetic regulation of neuron-dependent induction of astroglial synaptic protein GLT1. Glia 2010;58(03):277–286. Doi: 10.1002/glia.20922
https://doi.org/10.1002/glia.20922...
When it comes to astrocytes, some studies point to important functions performed by the EVs secreted by them. One study, for example, established that astrocytes secrete ectosomes that are rich in cytokines and other pro-inflammatory molecules, suggesting the action of EVs as neuromodulators.1919 Hewett SJ, Jackman NA, Claycomb RJ. Interleukin-1β in Central Nervous System Injury and Repair. Eur J Neurodegener Dis 2012;1 (02):195–211 As mentioned earlier, oligodendrocytes also secrete extracellular vesicles. Previous studies have proven that these cells secrete EVs which provide neurons with structural and trophic support.1414 Krämer-Albers EM, Bretz N, Tenzer S, et al. Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: Trophic support for axons? Proteomics Clin Appl 2007;1 (11):1446–1461. Doi: 10.1002/prca.200700522
https://doi.org/10.1002/prca.200700522...
,2020 Bakhti M, Winter C, Simons M. Inhibition of myelin membrane sheath formation by oligodendrocyte-derived exosome-like vesicles. J Biol Chem 2011;286(01):787–796. Doi: 10.1074/jbc.M110.190009
https://doi.org/10.1074/jbc.M110.190009...
A specific study pointed out an interesting result about a possible function of the EVs secreted by oligodendrocytes.2121 Fröhlich D, Kuo WP, Frühbeis C, et al. Multifaceted effects of oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal transduction and gene regulation. Philos Trans R Soc Lond B Biol Sci 2014;369(1652):20130510. Doi: 10.1098/rstb.2013.0510
https://doi.org/10.1098/rstb.2013.0510...
In this work it was found that primary rat neurons cultured in a culture medium rich in oligodendrocyte-derived EVs had more active metabolism in ischemic situations (lack of glucose and oxygen) than those not grown in this medium. This result is probably due to the transfer of the enzymes superoxide dismutase and catalase to the neurons via oligodendrocyte-derived EVs (ODEVs). In addition, one study performed a proteomic analysis of ODEVs.1414 Krämer-Albers EM, Bretz N, Tenzer S, et al. Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: Trophic support for axons? Proteomics Clin Appl 2007;1 (11):1446–1461. Doi: 10.1002/prca.200700522
https://doi.org/10.1002/prca.200700522...
In this analysis, the presence of important proteins for myelin formation was found, such as myelin basic protein and myelin oligodendrocyte glycoprotein, suggesting that ODEVs may play a role in the regulation of myelin sheath formation. Extrapolating their physiological functions, several studies point out that EVs may be related to the pathogenesis of various diseases, contributing to them, or acting as a reflection of cellular stress and/or injury, which allows their use as biomarkers.2222 Hill AF. Extracellular Vesicles and Neurodegenerative Diseases. J Neurosci 2019;39(47):9269–9273. Doi: 10.1523/JNEUROSCI.0147-18.2019
https://doi.org/10.1523/JNEUROSCI.0147-1...
Among the diseases in which it is thought that EVs may play an important role, one of the most researched is cancer. Studies have shown that the metastasis of tumor cells is preceded by the elimination of EV's by tumor cells and that these EV's would have the function of preparing the distant tissue for metastasis to be successful.2323 Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res 2011;71(11):3792–3801. Doi: 10.1158/0008-5472.CAN-10-4455
https://doi.org/10.1158/0008-5472.CAN-10...
,2424 Peinado H, Alečković M, Lavotshkin S, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 2012;18(06):883–891. Doi: 10.1038/nm.2753 Erratum in: NatMed. 2016 Dec 6;22(12):1502. PMID: 22635005; PMCID: PMC3645291
https://doi.org/10.1038/nm.2753...

EXTRACELLULAR VESICLES AS BIOMARKERS IN ALZHEIMER'S DISEASE

The role of EVs in the pathophysiology of neurodegenerative diseases such as Alzheimer's, Parkinson's, and prion diseases has been the subject of recent studies, due to the fact that these diseases share a similar mechanism, in which there is misfolding of certain proteins, their deposition and the subsequent dissemination to specific regions of the CNS. Since EVs play a role in cell signaling, it is thought that they may be a vehicle for the transmission of the misfolded proteins, thus contributing to the pathogenesis of the above diseases.2525 Braak H, Rüb U, Gai WP, Del Tredici K. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural Transm (Vienna) 2003;110(05):517–536. Doi: 10.1007/s00702-002-0808-2
https://doi.org/10.1007/s00702-002-0808-...

Alzheimer's disease (AD) is the most common neurodegenerative disease affecting the CNS, leading to neuronal death. When the clinical diagnosis is made, the disease is quite advanced and a large neuronal loss is already established, making the treatment possibilities not so effective. It is in this context that EVs may become important: recent studies indicate the possibility of using them as early diagnostics biomarkers of AD, enabling a more effective treatment in individuals diagnosed at earlier stages.2626 Ausó E, Gómez-Vicente V, Esquiva G. Biomarkers for Alzheimer’s Disease Early Diagnosis. J Pers Med 2020;10(03):114. Doi: 10.3390/jpm10030114
https://doi.org/10.3390/jpm10030114...
The pathophysiological changes in AD are accompanied by changes in both the quantity and composition of neuron-derived EVs (NDEVs), neuron precursor cells, and astrocytes.2727 Musunuri S, Khoonsari PE, Mikus M, et al. Increased Levels of Extracellular Microvesicle Markers and Decreased Levels of Endocytic/Exocytic Proteins in the Alzheimer’s Disease Brain. J Alzheimers Dis 2016;54(04):1671–1686. Doi: 10.3233/JAD-160271
https://doi.org/10.3233/JAD-160271...
2828 Badhwar A, Haqqani AS. Biomarker potential of brain-secreted extracellular vesicles in blood in Alzheimer’s disease. Alzheimers Dement (Amst) 2020;12(01):e12001. Doi: 10.1002/dad2.12001
https://doi.org/10.1002/dad2.12001...

TAU AND Aß

It has been shown that alterations in the neural synapse process modify the secretion of EVs vesicles by neurons.2929 Lugli G, Cohen AM, Bennett DA, et al. Plasma Exosomal miRNAs in Persons with and without Alzheimer Disease: Altered Expression and Prospects for Biomarkers. PLoS One 2015;10(10):e0139233. Doi: 10.1371/journal.pone.0139233
https://doi.org/10.1371/journal.pone.013...
,3030 Chivet M, Hemming F, Pernet-Gallay K, Fraboulet S, Sadoul R. Emerging role of neuronal exosomes in the central nervous system. Front Physiol 2012;3:145. Doi: 10.3389/fphys.2012.00145
https://doi.org/10.3389/fphys.2012.00145...
Furthermore, the presence of the most predisposing allele to AD, the APOE-ε4 allele, is related to decreased secretion of brain-derived EVs.3131 Peng KY, Pérez-González R, Alldred MJ, et al. Apolipoprotein E4 genotype compromises brain exosome production. Brain 2019; 142(01):163–175. Doi: 10.1093/brain/awy289
https://doi.org/10.1093/brain/awy289...
Aß(Amyloid beta) and tau are protein forms known to be involved in the pathophysiology of AD.3232 Tran L, Ha-Duong T. Exploring the Alzheimer amyloid-β peptide conformational ensemble: A review of molecular dynamics approaches. Peptides 2015;69:86–91. Doi: 10.1016/j.peptides.2015.04.009
https://doi.org/10.1016/j.peptides.2015....
,3333 Eftekharzadeh B, Daigle JG, Kapinos LE, et al. Tau Protein Disrupts Nucleocytoplasmic Transport in Alzheimer’s Disease. Neuron 2018;99(05):925–940.e7. Doi: 10.1016/j.neuron.2018.07.039 Erratum in: Neuron. 2019 Jan 16;101(2):349. PMID: 30189209; PMCID: PMC6240334
https://doi.org/10.1016/j.neuron.2018.07...
Aß is an anomalous insoluble protein formed by the differential cleavage of amyloid precursor protein(APP) by the enzymes β-secretase and γ-secretase in the brain of AD patients. Such cleavage leads to the formation of insoluble peptide fragments (Aß40/Aß42) that undergo oligomerization and subsequent polymerization in the synaptic environment, generating deleterious effects, such as alterations in calcium metabolism and energy metabolism, increased oxidative stress, and ion channel blockade, which contribute to the neuronal death seen in the disease.3434 Chen JX, Yan SS. Role of mitochondrial amyloid-beta in Alzheimer’s disease. J Alzheimers Dis 2010;20(Suppl 2):S569–S578. Doi: 10.3233/JAD-2010-100357
https://doi.org/10.3233/JAD-2010-100357...
,3535 Crews L, Masliah E.Molecular mechanisms of neurodegeneration in Alzheimer’s disease. Hum Mol Genet 2010;19(R1):R12–R20. Doi: 10.1093/hmg/ddq160
https://doi.org/10.1093/hmg/ddq160...
Another toxic effect of extracellular amyloid plaque formation is the increase in the pool of protein kinases, such as Glycogen Synthase kinase 3 (GSK3) and Cyclin-dependent kinase 5 (CDK5). These proteins are responsible for the hyperphosphorylation of tau protein fragments, leading to their aggregation and consequent formation of intracellular neurofibrillary tangles(NFTs).3333 Eftekharzadeh B, Daigle JG, Kapinos LE, et al. Tau Protein Disrupts Nucleocytoplasmic Transport in Alzheimer’s Disease. Neuron 2018;99(05):925–940.e7. Doi: 10.1016/j.neuron.2018.07.039 Erratum in: Neuron. 2019 Jan 16;101(2):349. PMID: 30189209; PMCID: PMC6240334
https://doi.org/10.1016/j.neuron.2018.07...
Since tau protein, in association with tubulin, is essential for microtubule homeostasis,3636 Claeysen S, Cochet M, Donneger R, Dumuis A, Bockaert J, Giannoni P. Alzheimer culprits: cellular crossroads and interplay. Cell Signal 2012;24(09):1831–1840. Doi: 10.1016/j.cellsig.2012.05.008
https://doi.org/10.1016/j.cellsig.2012.0...
it is reasonable to infer that interneuronal communication will be affected in the case of NFT formation, which also favors neuronal loss.3737 Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative tauopathies. Annu Rev Neurosci 2001;24:1121–1159. Doi: 10.1146/annurev.neuro.24.1.1121
https://doi.org/10.1146/annurev.neuro.24...
There is accumulated evidence pointing towards the use of both of this proteins (Aß and tau) as biomarkers of Alzheimer's Disease. More specifically in cerebrospinal fluid (CSF), it is thought that low levels of Aß42 are found due to increased deposition of Aß plaque in the encephalon, whereas a high level of tau and p-tau indicates a high degree of tau protein disturbances.3838 Jack CR Jr, Bennett DA, Blennow K, et al; Contributors. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement 2018;14(04):535–562. Doi: 10.1016/j.jalz.2018.02.018
https://doi.org/10.1016/j.jalz.2018.02.0...

39 Jack CR Jr, Bennett DA, Blennow K, et al. A/T/N: An unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology 2016;87(05):539–547. Doi: 10.1212/WNL.0000000000002923
https://doi.org/10.1212/WNL.000000000000...

40 Chen Z, Mengel D, Keshavan A, et al. Learnings about the complexity of extracellular tau aid development of a blood-based screen for Alzheimer’s disease. Alzheimers Dement 2019;15(03): 487–496. Doi: 10.1016/j.jalz.2018.09.010
https://doi.org/10.1016/j.jalz.2018.09.0...
-4141 Sato C, Barthélemy NR, Mawuenyega KG, et al. Tau Kinetics in Neurons and the Human Central Nervous System. Neuron 2018; 97(06):1284–1298.e7. Doi: 10.1016/j.neuron.2018.02.015 Erratum in: Neuron. 2018 May 16;98 (4):861–864. PMID: 29566794; PMCID: PMC6137722
https://doi.org/10.1016/j.neuron.2018.02...
In a murine model of AD, tau protein was shown to spread through the secretion of exosomes, especially by microglia cells, which was postulated after it was discovered that the depletion of these cells decreased such spread, suggesting that inhibition of EV secretion was beneficial in decreasing the spread of defective tau.4242 Asai H, Ikezu S, Tsunoda S, et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci 2015;18(11):1584–1593. Doi: 10.1038/nn.4132
https://doi.org/10.1038/nn.4132...
Furthermore, studies point to the possibility that EV transport of Aß protein in its oligomeric and neurotoxic form through neurons, which is due to the discovery that EVs can carry such protein from brain tissue analysis. These EVs were shown to be deleterious to the primary culture of neurons, demonstrating, in vitro, a possible action of EVs in the pathogenesis of the disease, through the transmission of Aß in its neurotoxic form through the CNS.4343 Sardar Sinha M, Ansell-Schultz A, Civitelli L, et al. Alzheimer’s disease pathology propagation by exosomes containing toxic amyloid-beta oligomers. Acta Neuropathol 2018;136(01): 41–56. Doi: 10.1007/s00401-018-1868-1
https://doi.org/10.1007/s00401-018-1868-...

Several studies investigating the levels of Aß42 in neuron-derived EVs isolated from plasma have reported increased amounts of this protein in individuals with AD and mild cognitive impairment (MCI) compared to healthy controls.4444 Jia L, Qiu Q, Zhang H, et al. Concordance between the assessment of Aβ42, T-tau, and P-T181-tau in peripheral blood neuronalderived exosomes and cerebrospinal fluid. Alzheimers Dement 2019;15(08):1071–1080. Doi: 10.1016/j.jalz.2019.05.002
https://doi.org/10.1016/j.jalz.2019.05.0...

45 Winston CN, Goetzl EJ, Baker LD, VitielloMV, Rissman RA. Growth Hormone-Releasing Hormone Modulation of Neuronal Exosome Biomarkers inMild Cognitive Impairment. J Alzheimers Dis 2018; 66(03):971–981. Doi: 10.3233/JAD-180302
https://doi.org/10.3233/JAD-180302...

46 Winston CN, Goetzl EJ, Akers JC, et al. Prediction of conversion from mild cognitive impairment to dementia with neuronally derived blood exosome protein profile. Alzheimers Dement (Amst) 2016;3:63–72. Doi: 10.1016/j.dadm.2016.04.001
https://doi.org/10.1016/j.dadm.2016.04.0...
-4747 Fiandaca MS, Kapogiannis D, Mapstone M, et al. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimers Dement 2015;11(06):600–7.e1. Doi: 10.1016/j.jalz.2014.06.008
https://doi.org/10.1016/j.jalz.2014.06.0...
One study in particular proved that this increase was present up to 10 years prior to AD diagnosis, as well as that these levels increased as the disease progressed.4747 Fiandaca MS, Kapogiannis D, Mapstone M, et al. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimers Dement 2015;11(06):600–7.e1. Doi: 10.1016/j.jalz.2014.06.008
https://doi.org/10.1016/j.jalz.2014.06.0...
This finding indicates that analysis of Aß42 levels in NBEVs isolated from plasma can serve as both an early biomarker, as well as a biomarker of AD progression. In this case, the use of plasma as the medium for EV extraction is preferred over CSF because the latter method is more invasive, as well as because a recent study has found that the number of EVs in plasma exceeds that found in cerebrospinal fluid.4848 Eitan E, Hutchison ER, Marosi K, et al. Extracellular Vesicle-Associated Aβ Mediates Trans-Neuronal Bioenergetic and Ca2þ-Handling Deficits in Alzheimer’s Disease Models. NPJ Aging Mech Dis 2016;2:16019. Doi: 10.1038/npjamd.2016.19
https://doi.org/10.1038/npjamd.2016.19...

Regarding the analysis of the presence of tau protein - another important component of Alzheirmer's pathophysiology - within neuron-derived EVs in AD, there is not enough conclusive evidence so far. Some studies pointed to high levels of p-tau in individuals with AD-associated dementia compared with controls, levels which reached a plateau about 10 years before clinical diagnosis,4444 Jia L, Qiu Q, Zhang H, et al. Concordance between the assessment of Aβ42, T-tau, and P-T181-tau in peripheral blood neuronalderived exosomes and cerebrospinal fluid. Alzheimers Dement 2019;15(08):1071–1080. Doi: 10.1016/j.jalz.2019.05.002
https://doi.org/10.1016/j.jalz.2019.05.0...
,4646 Winston CN, Goetzl EJ, Akers JC, et al. Prediction of conversion from mild cognitive impairment to dementia with neuronally derived blood exosome protein profile. Alzheimers Dement (Amst) 2016;3:63–72. Doi: 10.1016/j.dadm.2016.04.001
https://doi.org/10.1016/j.dadm.2016.04.0...
,4747 Fiandaca MS, Kapogiannis D, Mapstone M, et al. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimers Dement 2015;11(06):600–7.e1. Doi: 10.1016/j.jalz.2014.06.008
https://doi.org/10.1016/j.jalz.2014.06.0...
which indicates that p-tau analysis may be less efficient than Aß42 analysis with respect to the investigation of disease progression. On the other hand, three studies found no significant difference in the amount of p-tau in NDEVs in AD.4545 Winston CN, Goetzl EJ, Baker LD, VitielloMV, Rissman RA. Growth Hormone-Releasing Hormone Modulation of Neuronal Exosome Biomarkers inMild Cognitive Impairment. J Alzheimers Dis 2018; 66(03):971–981. Doi: 10.3233/JAD-180302
https://doi.org/10.3233/JAD-180302...
,4949 Shi M, Kovac A, Korff A, et al. CNS tau efflux via exosomes is likely increased in Parkinson’s disease but not in Alzheimer’s disease. Alzheimers Dement 2016;12(11):1125–1131,5050 Guix FX, Corbett GT, Cha DJ, et al. Detection of aggregationcompetent tau in neuron-derived extracellular vesicles. Int J Mol Sci 2018;19(03):663

NDEVs CONTAINING OTHER PROTEINS INVOLVED IN AD PATHOGENESIS

Tau and Aß are not the only proteins found in neuron-derived EVs that can be used as biomarkers for Alzheimer's. One study demonstrated that the levels of some synaptic proteins such as neurogranin, synaptotagmin, synaptopodin, and synaptophysin, were reduced in the EVs isolated from the plasma of individuals with AD. However, these proteins are also reduced in individuals with MCI and Parkinson's, so that their selectivity is not as high.5151 Goetzl EJ, Kapogiannis D, Schwartz JB, et al. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J 2016;30(12):4141–4148. Doi: 10.1096/fj.201600816R
https://doi.org/10.1096/fj.201600816R...
SNAP-25 and synapsin 1 proteins also have reduced levels in NDEVs isolated from the blood of AD patients compared to control subjects.5151 Goetzl EJ, Kapogiannis D, Schwartz JB, et al. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J 2016;30(12):4141–4148. Doi: 10.1096/fj.201600816R
https://doi.org/10.1096/fj.201600816R...
,5252 Agliardi C, Guerini FR, Zanzottera M, Bianchi A, Nemni R, ClericiM. SNAP-25 in SerumIs Carried by Exosomes of Neuronal Origin and Is a Potential Biomarker of Alzheimer’s Disease. Mol Neurobiol 2019;56(08):5792–5798. Doi: 10.1007/s12035-019-1501-x
https://doi.org/10.1007/s12035-019-1501-...
One study compared the levels of pre(neuronal pentraxin 2, neurexin 2 alpha) and postsynaptic proteins (GluA4-containing glutamate receptor, neuroligin 1) in NBEVs of AD subjects and normal subjects.5353 Goetzl EJ, Abner EL, Jicha GA, Kapogiannis D, Schwartz JB. Declining levels of functionally specialized synaptic proteins in plasma neuronal exosomes with progression of Alzheimer’s disease. FASEB J 2018;32(02):888–893. Doi: 10.1096/fj.201700731R
https://doi.org/10.1096/fj.201700731R...
As a result, the levels of the four aforementioned proteins were reduced in NDEVs from AD patients, and the levels of the postsynaptic proteins were directly related to cognitive loss. Also in this study, a longitudinal design was conducted, in which it was found that reductions in the levels of these proteins, except for neuronal pentraxin 2, were already observed 6-11 years before diagnosis, and that this was directly proportional to disease progression, indicating a possible use of these proteins as both early biomarkers and biomarkers of AD progression.

Furthermore, insulin metabolism-related proteins are found in NDEVs. Their use as biomarkers of AD has been recently proposed, based on the finding that increased levels of phospho-Ser312-IRS1 and decreased phospho-panTyr-IRS-1 were found in individuals with AD.5454 Mullins RJ, Mustapic M, Goetzl EJ, Kapogiannis D. Exosomal biomarkers of brain insulin resistance associated with regional atrophy in Alzheimer’s disease. Hum Brain Mapp 2017;38(04): 1933–1940. Doi: 10.1002/hbm.23494
https://doi.org/10.1002/hbm.23494...
,5555 Kapogiannis D, Boxer A, Schwartz JB, et al. Dysfunctionally phosphorylated type 1 insulin receptor substrate in neural-derived blood exosomes of preclinical Alzheimer’s disease. FASEB J 2015;29(02):589–596. Doi: 10.1096/fj.14-262048
https://doi.org/10.1096/fj.14-262048...
Lysosomal proteins found in NDEVs have also been considered as possible early biomarkers of AD. In the study of Goetzl, et al. 2015,5656 Goetzl EJ, Boxer A, Schwartz JB, et al. Altered lysosomal proteins in neural-derived plasma exosomes in preclinical Alzheimer disease. Neurology 2015;85(01):40–47 levels of cathepsin D, a lysosome-associated membrane protein, and ubiquitinated proteins were increased, whereas heat-shock protein-70 levels were decreased in individuals in the preclinical and clinical stages of AD.

EVs FROM OTHER CELL TYPES

As previously stated, it is possible to purify EVs derived from several CNS cells, not only neurons. One study analyzed the content of EVs derived from a specific type of neuron precursor cell: the CSPG4 cells (Chondroitin sulfate proteoglycan). These cells have the function of secreting substances that are determinants for the growth and survival of neurons. In this sense, EVs were found to have significantly lower levels of four neurotrophic factors (hepatocyte growth factor, fibroblast growth factors 2 and 13, and type 1 insulin-like growth factor) in AD patients in preclinical phase, exposing the possibility of using these proteins as early biomarkers of AD.5757 Goetzl EJ, Nogueras-Ortiz C, Mustapic M, et al. Deficient neurotrophic factors of CSPG4-type neural cell exosomes in Alzheimer disease. FASEB J 2019;33(01):231–238 Astrocyte-derived EVs also have potential biomarkers for AD. One study5858 Goetzl EJ, Schwartz JB, Abner EL, Jicha GA, Kapogiannis D. High complement levels in astrocyte-derived exosomes of Alzheimer disease. Ann Neurol 2018;83(03):544–552 found elevated levels of BACE1 (a protein that initiates pathological APP cleavage in Alzheimer's pathophysiology), sAPPß and complement proteins, while another study found low levels of glia-derived neurotrophic factor (GNDF) in individuals with AD.5959 Goetzl EJ, Mustapic M, Kapogiannis D, et al. Cargo proteins of plasma astrocyte-derived exosomes in Alzheimer’s disease. FASEB J 2016;30(11):3853–3859. Doi: 10.1096/fj.201600756R
https://doi.org/10.1096/fj.201600756R...

ANALYSIS OF EV CARGO

The analyses of the genetic content in EVs may be an important way of obtaining biomarkers for AD as well as in several other disease conditions. It has been shown that exosomes carry mRNA and microRNAs (miRNAs) in addition to proteins and other macromolecules,6060 Pant S, Hilton H, Burczynski ME. The multifaceted exosome: biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker opportunities. Biochem Pharmacol 2012;83(11):1484–1494 and abnormalities of miRNA expression have been found in exosomes from individuals with AD.6161 Burgos K, Malenica I, Metpally R, et al. Profiles of extracellular miRNA in cerebrospinal fluid and serum from patients with Alzheimer’s and Parkinson’s diseases correlate with disease status and features of pathology. PLoS One 2014;9(05):e94839 Some studies have already demonstrated differences in the expression of various types of miRNAs in exosomes purified from the plasma or serum of individuals with AD and MCI.2929 Lugli G, Cohen AM, Bennett DA, et al. Plasma Exosomal miRNAs in Persons with and without Alzheimer Disease: Altered Expression and Prospects for Biomarkers. PLoS One 2015;10(10):e0139233. Doi: 10.1371/journal.pone.0139233
https://doi.org/10.1371/journal.pone.013...
,6262 Liu CG, Song J, Zhang YQ,Wang PC. MicroRNA-193b is a regulator of amyloid precursor protein in the blood and cerebrospinal fluid derived exosomal microRNA-193b is a biomarker of Alzheimer’s disease. Mol Med Rep 2014;10(05):2395–2400

63 Cheng L, Doecke JD, Sharples RA, et al; Australian Imaging, Biomarkers and Lifestyle (AIBL) Research Group. Prognostic serum miRNA biomarkers associated with Alzheimer’s disease shows concordance with neuropsychological and neuroimaging assessment. Mol Psychiatry 2015;20(10):1188–1196
-6464 Yang TT, Liu CG, Gao SC, Zhang Y, Wang PC. The Serum Exosome Derived MicroRNA-135a, -193b, and -384 Were Potential Alzheimer’s Disease Biomarkers. Biomed Environ Sci 2018;31(02): 87–96 As an example of this, studies have found that the expression of the exosome-associated miRNAs miR-342-3p,miR-125a-5p, miR-125b-5p, and miR-451a, was significantly lower in individuals with AD, and a correlation can be made between the differences in expression and the level of cognitive defect.2929 Lugli G, Cohen AM, Bennett DA, et al. Plasma Exosomal miRNAs in Persons with and without Alzheimer Disease: Altered Expression and Prospects for Biomarkers. PLoS One 2015;10(10):e0139233. Doi: 10.1371/journal.pone.0139233
https://doi.org/10.1371/journal.pone.013...
,6565 Rani A, O’Shea A, Ianov L, Cohen RA, Woods AJ, Foster TC. miRNA in circulating microvesicles as biomarkers for age-related cognitive decline. Front Aging Neurosci 2017;9:323 Interestingly, a study6464 Yang TT, Liu CG, Gao SC, Zhang Y, Wang PC. The Serum Exosome Derived MicroRNA-135a, -193b, and -384 Were Potential Alzheimer’s Disease Biomarkers. Biomed Environ Sci 2018;31(02): 87–96 pointed out that the combined analysis of the exosomal expression of miRNAs miR-135a, miR-193b, and miR-384, related to the modulation of APP and/or BACE1 expression, is a good biomarker of early AD. In this sense, one may suggest that a combined analysis of several molecules may be more valuable than the isolated analysis of possible biomarkers for a possible early diagnosis.

A recent study analyzed the content of small nuclear-RNAs(snRNAs) in plasma-derived EV's and their use as a biomarker of AD.6666 Fitz NF, Wang J, Kamboh MI, Koldamova R, Lefterov I. Small nucleolar RNAs in plasma extracellular vesicles and their discriminatory power as diagnostic biomarkers of Alzheimer’s disease. Neurobiol Dis 2021;159:105481. Doi: 10.1016/j.nbd.2021.105481
https://doi.org/10.1016/j.nbd.2021.10548...
Levels of 4 snRNAs: miRNAs(microRNAs), snoRNAs (small-nucleolarRNAs), tRNAs (transferRNAs) and piRNAs (piwi-interactingRNAs) were analyzed via droplet-digital-PCR(ddPCR). The results showed that there was significantly different expression of two snoRNAs, SNORD115 and SNORD 116, both of which are predominantly expressed in the brain.6767 Cavaillé J Box C/D small nucleolar RNA genes and the Prader-Willi syndrome: a complex interplay.Wiley Interdiscip Rev RNA 2017; 8(04 Epub2017Mar13. Doi: 10.1002/wrna.1417
https://doi.org/10.1002/wrna.1417...

68 Chung MS, Langouët M, Chamberlain SJ, Carmichael GG. Prader-Willi syndrome: reflections on seminal studies and future therapies. Open Biol 2020;10(09):200195. Doi: 10.1098/rsob.200195
https://doi.org/10.1098/rsob.200195...

69 Driedonks TAP, van der Grein SG, Ariyurek Y, et al. Immune stimuli shape thesmall non-coding transcriptome of extracellular vesicles released by dendritic cells. Cell Mol Life Sci 2018;75(20): 3857–3875. Doi: 10.1007/s00018-018-2842-8
https://doi.org/10.1007/s00018-018-2842-...

70 Irimie AI, Zimta AA, Ciocan C, et al. The Unforeseen Non-Coding RNAs in Head and Neck Cancer. Genes (Basel) 2018;9(03):134.Doi: 10.3390/genes9030134
https://doi.org/10.3390/genes9030134...
-7171 Lässer C, Shelke GV, Yeri A, et al. Two distinct extracellular RNA signatures released by a single cell type identified by microarray and next-generation sequencing. RNA Biol 2017;14(01):58–72. Doi: 10.1080/15476286.2016.1249092
https://doi.org/10.1080/15476286.2016.12...
Such differential expression of these two snoRNAs in EVs isolated from the plasma of individuals, when analyzed combined, enabled the differentiation of AD from healthy controls(Area under the curve of 94.7%), higher than other biomarkers. Another paper7272 Kim KM, Meng Q, Perez de Acha O, et al. Mitochondrial RNA in Alzheimer’s Disease Circulating Extracellular Vesicles. Front Cell Dev Biol 2020;8:581882. Doi: 10.3389/fcell.2020.581882
https://doi.org/10.3389/fcell.2020.58188...
aimed to ascertain whether there are differences in mitochondrial RNA (mtRNA) content in EVs isolated from plasma between AD, MCI, and controls (age-matched) individuals. Using the RNA-seq technique, the researchers confirmed significantly elevated levels of certain mitochondrial RNAs (MT-ND1-6, MT-ND4L, MT-ATP6, MTATP8, MT-CYTB, MT-CO1, MT-CO2, MT-CO3 mRNAs, and MT-RNR1 rRNA) in EVs isolated from the plasma of AD and MCI. The researchers proposed that since the pathophysiological mechanisms of AD cause mitochondrial damage,7373 Beal MF. Mitochondrial dysfunction in neurodegenerative diseases. Biochim Biophys Acta 1998;1366(1-2):211–223. Doi: 10.1016/s0005-2728(98)00114-5
https://doi.org/10.1016/s0005-2728(98)00...
,7474 Beal MF. Oxidative damage as an early marker of Alzheimer’s disease andmild cognitive impairment. Neurobiol Aging 2005;26 (05):585–586. Doi: 10.1016/j.neurobiolaging.2004.09.022
https://doi.org/10.1016/j.neurobiolaging...
there is secretion of mitochondrial components through EVs, opening up the possibility for its use as a diagnostic and prognostic biomarker. It is important to mention that in this study EVs were not isolated from CNS cells, since the researchers pointed out that the isolation methods resulted in a very low pool of EVs, which would impair the quality of the study. However, the researchers performed an experiment in which they cultured several CNS cells (microglia, astrocytes, and neurons), imposed on them conditions similar to those seen in AD (Aß42 and ROS), and found EVs containing high levels of mt-RNAs compared to control cultures.

EVs contain, besides proteins and genetic material, lipids, both in the constitution of their membrane and cargo, so it may be possible to use the dosage of these lipids as a possible biomarker of AD. A specific study7575 Su H, Rustam YH, Masters CL, et al. Characterization of brainderived extracellular vesicle lipids in Alzheimer’s disease. J Extracell Vesicles 2021;10(07):e12089. Doi: 10.1002/jev2.12089
https://doi.org/10.1002/jev2.12089...
aimed to investigate whether there are differences in the expression of lipids in Brain-Derived Extracellular Vesicles(BEDVs) between AD and age-matched control individuals, more specifically coming from the frontal cortex tissue of the participants. The study found increased plasmalogen glycerophosphoethanolamine and decreased polyunsaturated fatty acyl-containing lipids in AD BDEVs. All the above information regarding the molecules cited in this article and their potential as biomarkers is summarized in Table 1.

Table 1
Main molecules and findings

CONCLUSION AND PERSPECTIVES

Extracellular vesicles (EVs) correspond to entities composed of a phospholipid bilayer, formed from endosomes or from the evagination of the plasma membrane of cells and their classification is still a topic under discussion.11 Théry C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018;7(01):1535750. Doi: 10.1080/20013078.2018.1535750
https://doi.org/10.1080/20013078.2018.15...

2 Théry C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol 2009;9(08):581–593. Doi: 10.1038/nri2567
https://doi.org/10.1038/nri2567...

3 Denzer K, Kleijmeer MJ, Heijnen HF, Stoorvogel W, Geuze HJ. Exosome: from internal vesicle of the multivesicular body to intercellular signaling device. J Cell Sci 2000;113(Pt 19): 3365–3374. Doi: 10.1242/jcs.113.19.3365
https://doi.org/10.1242/jcs.113.19.3365...
-44 György B, Szabó TG, Pásztói M, et al. Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles. Cell Mol Life Sci 2011;68(16):2667–2688. Doi: 10.1007/s00018-011-0689-3
https://doi.org/10.1007/s00018-011-0689-...
These vesicles carry within them different contents, such as genetic material, proteins, lipids, and carbohydrates, which come from the cells of origin of the vesicle, thus reflecting the state of such cells.55 Raposo G, StoorvogelW. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 2013;200(04):373–383. Doi: 10.1083/jcb.201211138
https://doi.org/10.1083/jcb.201211138...

6 Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function.Nat Rev Immunol 2002;2(08):569–579. Doi: 10.1038/nri855
https://doi.org/10.1038/nri855...
-77 Shah R, Patel T, Freedman JE. Circulating Extracellular Vesicles in Human Disease. N Engl J Med 2018;379(10):958–966. Doi: 10.1056/NEJMra1704286
https://doi.org/10.1056/NEJMra1704286...
EVs are already known to have diverse functions, with their participation in cell signaling being one of the best understood. However, their presence in systemic processes has already been verified, as well as in several pathologies, such as neurodegenerative diseases, Alzheimer's being the disease addressed by this article.2525 Braak H, Rüb U, Gai WP, Del Tredici K. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural Transm (Vienna) 2003;110(05):517–536. Doi: 10.1007/s00702-002-0808-2
https://doi.org/10.1007/s00702-002-0808-...
In this sense, several alterations have been verified in both the number and content of extracellular vesicles extracted from patients with Alzheimer's disease(AD), or in animal models. Because EV's can be isolated from various biofluids such as CSF, blood, urine, among others, their use as a biomarker is quite an exciting prospect, allowing better screening and treatment chances for the patients. The aim of the article, therefore, was to gather the previously published information about the use of EVs as biomarkers in AD. Although the information is promising, more studies are needed, especially longitudinal studies, to obtain a reliable pattern of change in the content and number of EVs, a pattern that can be used on a large scale and is very efficient in the early diagnosis of AD, in order to improve the quality of life of patients.

References

  • 1
    Théry C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018;7(01):1535750. Doi: 10.1080/20013078.2018.1535750
    » https://doi.org/10.1080/20013078.2018.1535750
  • 2
    Théry C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol 2009;9(08):581–593. Doi: 10.1038/nri2567
    » https://doi.org/10.1038/nri2567
  • 3
    Denzer K, Kleijmeer MJ, Heijnen HF, Stoorvogel W, Geuze HJ. Exosome: from internal vesicle of the multivesicular body to intercellular signaling device. J Cell Sci 2000;113(Pt 19): 3365–3374. Doi: 10.1242/jcs.113.19.3365
    » https://doi.org/10.1242/jcs.113.19.3365
  • 4
    György B, Szabó TG, Pásztói M, et al. Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles. Cell Mol Life Sci 2011;68(16):2667–2688. Doi: 10.1007/s00018-011-0689-3
    » https://doi.org/10.1007/s00018-011-0689-3
  • 5
    Raposo G, StoorvogelW. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 2013;200(04):373–383. Doi: 10.1083/jcb.201211138
    » https://doi.org/10.1083/jcb.201211138
  • 6
    Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function.Nat Rev Immunol 2002;2(08):569–579. Doi: 10.1038/nri855
    » https://doi.org/10.1038/nri855
  • 7
    Shah R, Patel T, Freedman JE. Circulating Extracellular Vesicles in Human Disease. N Engl J Med 2018;379(10):958–966. Doi: 10.1056/NEJMra1704286
    » https://doi.org/10.1056/NEJMra1704286
  • 8
    Raposo G, Nijman HW, StoorvogelW, et al. B lymphocytes secrete antigen-presenting vesicles. J Exp Med 1996;183(03):1161–1172
  • 9
    Robbins PD, Dorronsoro A, Booker CN. Regulation of chronic inflammatory and immune processes by extracellular vesicles. J Clin Invest 2016;126(04):1173–1180
  • 10
    Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol 2014;30:255–289. Doi: 10.1146/annurevcellbio-101512-122326
    » https://doi.org/10.1146/annurev-cellbio-101512-122326
  • 11
    Fauré J, Lachenal G, Court M, et al. Exosomes are released by cultured cortical neurones. Mol Cell Neurosci 2006;31(04): 642–648. Doi: 10.1016/j.mcn.2005.12.003
    » https://doi.org/10.1016/j.mcn.2005.12.003
  • 12
    Morel L, Regan M, Higashimori H, et al. Neuronal exosomal miRNA-dependent translational regulation of astroglial glutamate transporter GLT1. J Biol Chem 2013;288(10):7105–7116. Doi: 10.1074/jbc.M112.410944
    » https://doi.org/10.1074/jbc.M112.410944
  • 13
    Potolicchio I, Carven GJ, Xu X, et al. Proteomic analysis of microglia-derived exosomes: metabolic role of the aminopeptidase CD13 in neuropeptide catabolism. J Immunol 2005;175(04): 2237–2243. Doi: 10.4049/jimmunol.175.4.2237
    » https://doi.org/10.4049/jimmunol.175.4.2237
  • 14
    Krämer-Albers EM, Bretz N, Tenzer S, et al. Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: Trophic support for axons? Proteomics Clin Appl 2007;1 (11):1446–1461. Doi: 10.1002/prca.200700522
    » https://doi.org/10.1002/prca.200700522
  • 15
    Saeedi S, Israel S, Nagy C, Turecki G. The emerging role of exosomes in mental disorders. Transl Psychiatry 2019;9(01): 122. Doi: 10.1038/s41398-019-0459-9
    » https://doi.org/10.1038/s41398-019-0459-9
  • 16
    Chivet M, Javalet C, Hemming F, et al. Exosomes as a novel way of interneuronal communication. Biochem Soc Trans 2013;41(01): 241–244. Doi: 10.1042/BST20120266
    » https://doi.org/10.1042/BST20120266
  • 17
    Goldie BJ, Dun MD, Lin M, et al. Activity-associated miRNA are packaged in Map1b-enriched exosomes released from depolarized neurons. Nucleic Acids Res 2014;42(14):9195–9208. Doi: 10.1093/nar/gku594
    » https://doi.org/10.1093/nar/gku594
  • 18
    Yang Y, Gozen O, Vidensky S, RobinsonMB, Rothstein JD. Epigenetic regulation of neuron-dependent induction of astroglial synaptic protein GLT1. Glia 2010;58(03):277–286. Doi: 10.1002/glia.20922
    » https://doi.org/10.1002/glia.20922
  • 19
    Hewett SJ, Jackman NA, Claycomb RJ. Interleukin-1β in Central Nervous System Injury and Repair. Eur J Neurodegener Dis 2012;1 (02):195–211
  • 20
    Bakhti M, Winter C, Simons M. Inhibition of myelin membrane sheath formation by oligodendrocyte-derived exosome-like vesicles. J Biol Chem 2011;286(01):787–796. Doi: 10.1074/jbc.M110.190009
    » https://doi.org/10.1074/jbc.M110.190009
  • 21
    Fröhlich D, Kuo WP, Frühbeis C, et al. Multifaceted effects of oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal transduction and gene regulation. Philos Trans R Soc Lond B Biol Sci 2014;369(1652):20130510. Doi: 10.1098/rstb.2013.0510
    » https://doi.org/10.1098/rstb.2013.0510
  • 22
    Hill AF. Extracellular Vesicles and Neurodegenerative Diseases. J Neurosci 2019;39(47):9269–9273. Doi: 10.1523/JNEUROSCI.0147-18.2019
    » https://doi.org/10.1523/JNEUROSCI.0147-18.2019
  • 23
    Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res 2011;71(11):3792–3801. Doi: 10.1158/0008-5472.CAN-10-4455
    » https://doi.org/10.1158/0008-5472.CAN-10-4455
  • 24
    Peinado H, Alečković M, Lavotshkin S, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 2012;18(06):883–891. Doi: 10.1038/nm.2753 Erratum in: NatMed. 2016 Dec 6;22(12):1502. PMID: 22635005; PMCID: PMC3645291
    » https://doi.org/10.1038/nm.2753
  • 25
    Braak H, Rüb U, Gai WP, Del Tredici K. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural Transm (Vienna) 2003;110(05):517–536. Doi: 10.1007/s00702-002-0808-2
    » https://doi.org/10.1007/s00702-002-0808-2
  • 26
    Ausó E, Gómez-Vicente V, Esquiva G. Biomarkers for Alzheimer’s Disease Early Diagnosis. J Pers Med 2020;10(03):114. Doi: 10.3390/jpm10030114
    » https://doi.org/10.3390/jpm10030114
  • 27
    Musunuri S, Khoonsari PE, Mikus M, et al. Increased Levels of Extracellular Microvesicle Markers and Decreased Levels of Endocytic/Exocytic Proteins in the Alzheimer’s Disease Brain. J Alzheimers Dis 2016;54(04):1671–1686. Doi: 10.3233/JAD-160271
    » https://doi.org/10.3233/JAD-160271
  • 28
    Badhwar A, Haqqani AS. Biomarker potential of brain-secreted extracellular vesicles in blood in Alzheimer’s disease. Alzheimers Dement (Amst) 2020;12(01):e12001. Doi: 10.1002/dad2.12001
    » https://doi.org/10.1002/dad2.12001
  • 29
    Lugli G, Cohen AM, Bennett DA, et al. Plasma Exosomal miRNAs in Persons with and without Alzheimer Disease: Altered Expression and Prospects for Biomarkers. PLoS One 2015;10(10):e0139233. Doi: 10.1371/journal.pone.0139233
    » https://doi.org/10.1371/journal.pone.0139233
  • 30
    Chivet M, Hemming F, Pernet-Gallay K, Fraboulet S, Sadoul R. Emerging role of neuronal exosomes in the central nervous system. Front Physiol 2012;3:145. Doi: 10.3389/fphys.2012.00145
    » https://doi.org/10.3389/fphys.2012.00145
  • 31
    Peng KY, Pérez-González R, Alldred MJ, et al. Apolipoprotein E4 genotype compromises brain exosome production. Brain 2019; 142(01):163–175. Doi: 10.1093/brain/awy289
    » https://doi.org/10.1093/brain/awy289
  • 32
    Tran L, Ha-Duong T. Exploring the Alzheimer amyloid-β peptide conformational ensemble: A review of molecular dynamics approaches. Peptides 2015;69:86–91. Doi: 10.1016/j.peptides.2015.04.009
    » https://doi.org/10.1016/j.peptides.2015.04.009
  • 33
    Eftekharzadeh B, Daigle JG, Kapinos LE, et al. Tau Protein Disrupts Nucleocytoplasmic Transport in Alzheimer’s Disease. Neuron 2018;99(05):925–940.e7. Doi: 10.1016/j.neuron.2018.07.039 Erratum in: Neuron. 2019 Jan 16;101(2):349. PMID: 30189209; PMCID: PMC6240334
    » https://doi.org/10.1016/j.neuron.2018.07.039
  • 34
    Chen JX, Yan SS. Role of mitochondrial amyloid-beta in Alzheimer’s disease. J Alzheimers Dis 2010;20(Suppl 2):S569–S578. Doi: 10.3233/JAD-2010-100357
    » https://doi.org/10.3233/JAD-2010-100357
  • 35
    Crews L, Masliah E.Molecular mechanisms of neurodegeneration in Alzheimer’s disease. Hum Mol Genet 2010;19(R1):R12–R20. Doi: 10.1093/hmg/ddq160
    » https://doi.org/10.1093/hmg/ddq160
  • 36
    Claeysen S, Cochet M, Donneger R, Dumuis A, Bockaert J, Giannoni P. Alzheimer culprits: cellular crossroads and interplay. Cell Signal 2012;24(09):1831–1840. Doi: 10.1016/j.cellsig.2012.05.008
    » https://doi.org/10.1016/j.cellsig.2012.05.008
  • 37
    Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative tauopathies. Annu Rev Neurosci 2001;24:1121–1159. Doi: 10.1146/annurev.neuro.24.1.1121
    » https://doi.org/10.1146/annurev.neuro.24.1.1121
  • 38
    Jack CR Jr, Bennett DA, Blennow K, et al; Contributors. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement 2018;14(04):535–562. Doi: 10.1016/j.jalz.2018.02.018
    » https://doi.org/10.1016/j.jalz.2018.02.018
  • 39
    Jack CR Jr, Bennett DA, Blennow K, et al. A/T/N: An unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology 2016;87(05):539–547. Doi: 10.1212/WNL.0000000000002923
    » https://doi.org/10.1212/WNL.0000000000002923
  • 40
    Chen Z, Mengel D, Keshavan A, et al. Learnings about the complexity of extracellular tau aid development of a blood-based screen for Alzheimer’s disease. Alzheimers Dement 2019;15(03): 487–496. Doi: 10.1016/j.jalz.2018.09.010
    » https://doi.org/10.1016/j.jalz.2018.09.010
  • 41
    Sato C, Barthélemy NR, Mawuenyega KG, et al. Tau Kinetics in Neurons and the Human Central Nervous System. Neuron 2018; 97(06):1284–1298.e7. Doi: 10.1016/j.neuron.2018.02.015 Erratum in: Neuron. 2018 May 16;98 (4):861–864. PMID: 29566794; PMCID: PMC6137722
    » https://doi.org/10.1016/j.neuron.2018.02.015
  • 42
    Asai H, Ikezu S, Tsunoda S, et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci 2015;18(11):1584–1593. Doi: 10.1038/nn.4132
    » https://doi.org/10.1038/nn.4132
  • 43
    Sardar Sinha M, Ansell-Schultz A, Civitelli L, et al. Alzheimer’s disease pathology propagation by exosomes containing toxic amyloid-beta oligomers. Acta Neuropathol 2018;136(01): 41–56. Doi: 10.1007/s00401-018-1868-1
    » https://doi.org/10.1007/s00401-018-1868-1
  • 44
    Jia L, Qiu Q, Zhang H, et al. Concordance between the assessment of Aβ42, T-tau, and P-T181-tau in peripheral blood neuronalderived exosomes and cerebrospinal fluid. Alzheimers Dement 2019;15(08):1071–1080. Doi: 10.1016/j.jalz.2019.05.002
    » https://doi.org/10.1016/j.jalz.2019.05.002
  • 45
    Winston CN, Goetzl EJ, Baker LD, VitielloMV, Rissman RA. Growth Hormone-Releasing Hormone Modulation of Neuronal Exosome Biomarkers inMild Cognitive Impairment. J Alzheimers Dis 2018; 66(03):971–981. Doi: 10.3233/JAD-180302
    » https://doi.org/10.3233/JAD-180302
  • 46
    Winston CN, Goetzl EJ, Akers JC, et al. Prediction of conversion from mild cognitive impairment to dementia with neuronally derived blood exosome protein profile. Alzheimers Dement (Amst) 2016;3:63–72. Doi: 10.1016/j.dadm.2016.04.001
    » https://doi.org/10.1016/j.dadm.2016.04.001
  • 47
    Fiandaca MS, Kapogiannis D, Mapstone M, et al. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimers Dement 2015;11(06):600–7.e1. Doi: 10.1016/j.jalz.2014.06.008
    » https://doi.org/10.1016/j.jalz.2014.06.008
  • 48
    Eitan E, Hutchison ER, Marosi K, et al. Extracellular Vesicle-Associated Aβ Mediates Trans-Neuronal Bioenergetic and Ca2þ-Handling Deficits in Alzheimer’s Disease Models. NPJ Aging Mech Dis 2016;2:16019. Doi: 10.1038/npjamd.2016.19
    » https://doi.org/10.1038/npjamd.2016.19
  • 49
    Shi M, Kovac A, Korff A, et al. CNS tau efflux via exosomes is likely increased in Parkinson’s disease but not in Alzheimer’s disease. Alzheimers Dement 2016;12(11):1125–1131
  • 50
    Guix FX, Corbett GT, Cha DJ, et al. Detection of aggregationcompetent tau in neuron-derived extracellular vesicles. Int J Mol Sci 2018;19(03):663
  • 51
    Goetzl EJ, Kapogiannis D, Schwartz JB, et al. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J 2016;30(12):4141–4148. Doi: 10.1096/fj.201600816R
    » https://doi.org/10.1096/fj.201600816R
  • 52
    Agliardi C, Guerini FR, Zanzottera M, Bianchi A, Nemni R, ClericiM. SNAP-25 in SerumIs Carried by Exosomes of Neuronal Origin and Is a Potential Biomarker of Alzheimer’s Disease. Mol Neurobiol 2019;56(08):5792–5798. Doi: 10.1007/s12035-019-1501-x
    » https://doi.org/10.1007/s12035-019-1501-x
  • 53
    Goetzl EJ, Abner EL, Jicha GA, Kapogiannis D, Schwartz JB. Declining levels of functionally specialized synaptic proteins in plasma neuronal exosomes with progression of Alzheimer’s disease. FASEB J 2018;32(02):888–893. Doi: 10.1096/fj.201700731R
    » https://doi.org/10.1096/fj.201700731R
  • 54
    Mullins RJ, Mustapic M, Goetzl EJ, Kapogiannis D. Exosomal biomarkers of brain insulin resistance associated with regional atrophy in Alzheimer’s disease. Hum Brain Mapp 2017;38(04): 1933–1940. Doi: 10.1002/hbm.23494
    » https://doi.org/10.1002/hbm.23494
  • 55
    Kapogiannis D, Boxer A, Schwartz JB, et al. Dysfunctionally phosphorylated type 1 insulin receptor substrate in neural-derived blood exosomes of preclinical Alzheimer’s disease. FASEB J 2015;29(02):589–596. Doi: 10.1096/fj.14-262048
    » https://doi.org/10.1096/fj.14-262048
  • 56
    Goetzl EJ, Boxer A, Schwartz JB, et al. Altered lysosomal proteins in neural-derived plasma exosomes in preclinical Alzheimer disease. Neurology 2015;85(01):40–47
  • 57
    Goetzl EJ, Nogueras-Ortiz C, Mustapic M, et al. Deficient neurotrophic factors of CSPG4-type neural cell exosomes in Alzheimer disease. FASEB J 2019;33(01):231–238
  • 58
    Goetzl EJ, Schwartz JB, Abner EL, Jicha GA, Kapogiannis D. High complement levels in astrocyte-derived exosomes of Alzheimer disease. Ann Neurol 2018;83(03):544–552
  • 59
    Goetzl EJ, Mustapic M, Kapogiannis D, et al. Cargo proteins of plasma astrocyte-derived exosomes in Alzheimer’s disease. FASEB J 2016;30(11):3853–3859. Doi: 10.1096/fj.201600756R
    » https://doi.org/10.1096/fj.201600756R
  • 60
    Pant S, Hilton H, Burczynski ME. The multifaceted exosome: biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker opportunities. Biochem Pharmacol 2012;83(11):1484–1494
  • 61
    Burgos K, Malenica I, Metpally R, et al. Profiles of extracellular miRNA in cerebrospinal fluid and serum from patients with Alzheimer’s and Parkinson’s diseases correlate with disease status and features of pathology. PLoS One 2014;9(05):e94839
  • 62
    Liu CG, Song J, Zhang YQ,Wang PC. MicroRNA-193b is a regulator of amyloid precursor protein in the blood and cerebrospinal fluid derived exosomal microRNA-193b is a biomarker of Alzheimer’s disease. Mol Med Rep 2014;10(05):2395–2400
  • 63
    Cheng L, Doecke JD, Sharples RA, et al; Australian Imaging, Biomarkers and Lifestyle (AIBL) Research Group. Prognostic serum miRNA biomarkers associated with Alzheimer’s disease shows concordance with neuropsychological and neuroimaging assessment. Mol Psychiatry 2015;20(10):1188–1196
  • 64
    Yang TT, Liu CG, Gao SC, Zhang Y, Wang PC. The Serum Exosome Derived MicroRNA-135a, -193b, and -384 Were Potential Alzheimer’s Disease Biomarkers. Biomed Environ Sci 2018;31(02): 87–96
  • 65
    Rani A, O’Shea A, Ianov L, Cohen RA, Woods AJ, Foster TC. miRNA in circulating microvesicles as biomarkers for age-related cognitive decline. Front Aging Neurosci 2017;9:323
  • 66
    Fitz NF, Wang J, Kamboh MI, Koldamova R, Lefterov I. Small nucleolar RNAs in plasma extracellular vesicles and their discriminatory power as diagnostic biomarkers of Alzheimer’s disease. Neurobiol Dis 2021;159:105481. Doi: 10.1016/j.nbd.2021.105481
    » https://doi.org/10.1016/j.nbd.2021.105481
  • 67
    Cavaillé J Box C/D small nucleolar RNA genes and the Prader-Willi syndrome: a complex interplay.Wiley Interdiscip Rev RNA 2017; 8(04 Epub2017Mar13. Doi: 10.1002/wrna.1417
    » https://doi.org/10.1002/wrna.1417
  • 68
    Chung MS, Langouët M, Chamberlain SJ, Carmichael GG. Prader-Willi syndrome: reflections on seminal studies and future therapies. Open Biol 2020;10(09):200195. Doi: 10.1098/rsob.200195
    » https://doi.org/10.1098/rsob.200195
  • 69
    Driedonks TAP, van der Grein SG, Ariyurek Y, et al. Immune stimuli shape thesmall non-coding transcriptome of extracellular vesicles released by dendritic cells. Cell Mol Life Sci 2018;75(20): 3857–3875. Doi: 10.1007/s00018-018-2842-8
    » https://doi.org/10.1007/s00018-018-2842-8
  • 70
    Irimie AI, Zimta AA, Ciocan C, et al. The Unforeseen Non-Coding RNAs in Head and Neck Cancer. Genes (Basel) 2018;9(03):134.Doi: 10.3390/genes9030134
    » https://doi.org/10.3390/genes9030134
  • 71
    Lässer C, Shelke GV, Yeri A, et al. Two distinct extracellular RNA signatures released by a single cell type identified by microarray and next-generation sequencing. RNA Biol 2017;14(01):58–72. Doi: 10.1080/15476286.2016.1249092
    » https://doi.org/10.1080/15476286.2016.1249092
  • 72
    Kim KM, Meng Q, Perez de Acha O, et al. Mitochondrial RNA in Alzheimer’s Disease Circulating Extracellular Vesicles. Front Cell Dev Biol 2020;8:581882. Doi: 10.3389/fcell.2020.581882
    » https://doi.org/10.3389/fcell.2020.581882
  • 73
    Beal MF. Mitochondrial dysfunction in neurodegenerative diseases. Biochim Biophys Acta 1998;1366(1-2):211–223. Doi: 10.1016/s0005-2728(98)00114-5
    » https://doi.org/10.1016/s0005-2728(98)00114-5
  • 74
    Beal MF. Oxidative damage as an early marker of Alzheimer’s disease andmild cognitive impairment. Neurobiol Aging 2005;26 (05):585–586. Doi: 10.1016/j.neurobiolaging.2004.09.022
    » https://doi.org/10.1016/j.neurobiolaging.2004.09.022
  • 75
    Su H, Rustam YH, Masters CL, et al. Characterization of brainderived extracellular vesicle lipids in Alzheimer’s disease. J Extracell Vesicles 2021;10(07):e12089. Doi: 10.1002/jev2.12089
    » https://doi.org/10.1002/jev2.12089

Publication Dates

  • Publication in this collection
    05 Apr 2024
  • Date of issue
    2024

History

  • Received
    15 June 2023
  • Reviewed
    30 Oct 2023
  • Accepted
    01 Dec 2023
Academia Brasileira de Neurologia - ABNEURO R. Vergueiro, 1353 sl.1404 - Ed. Top Towers Offices Torre Norte, 04101-000 São Paulo SP Brazil, Tel.: +55 11 5084-9463 | +55 11 5083-3876 - São Paulo - SP - Brazil
E-mail: revista.arquivos@abneuro.org